Dendritic cell-specific delivery of Flt3L by coronavirus vectors secures induction of therapeutic antitumor immunity.

Efficacy of antitumor vaccination depends to a large extent on antigen targeting to dendritic cells (DCs). Here, we assessed antitumor immunity induced by attenuated coronavirus vectors which exclusively target DCs in vivo and express either lymphocyte- or DC-activating cytokines in combination with...

Full description

Bibliographic Details
Main Authors: Christian Perez-Shibayama, Cristina Gil-Cruz, Monika Nussbacher, Eva Allgäuer, Luisa Cervantes-Barragan, Roland Züst, Burkhard Ludewig
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2013-01-01
Series:PLoS ONE
Online Access:http://europepmc.org/articles/PMC3842931?pdf=render
id doaj-8e39703a017b4475a1f32c11b84963e9
record_format Article
spelling doaj-8e39703a017b4475a1f32c11b84963e92020-11-25T01:23:56ZengPublic Library of Science (PLoS)PLoS ONE1932-62032013-01-01811e8144210.1371/journal.pone.0081442Dendritic cell-specific delivery of Flt3L by coronavirus vectors secures induction of therapeutic antitumor immunity.Christian Perez-ShibayamaCristina Gil-CruzMonika NussbacherEva AllgäuerLuisa Cervantes-BarraganRoland ZüstBurkhard LudewigEfficacy of antitumor vaccination depends to a large extent on antigen targeting to dendritic cells (DCs). Here, we assessed antitumor immunity induced by attenuated coronavirus vectors which exclusively target DCs in vivo and express either lymphocyte- or DC-activating cytokines in combination with a GFP-tagged model antigen. Tracking of in vivo transduced DCs revealed that vectors encoding for Fms-like tyrosine kinase 3 ligand (Flt3L) exhibited a higher capacity to induce DC maturation compared to vectors delivering IL-2 or IL-15. Moreover, Flt3L vectors more efficiently induced tumor-specific CD8(+) T cells, expanded the epitope repertoire, and provided both prophylactic and therapeutic tumor immunity. In contrast, IL-2- or IL-15-encoding vectors showed a substantially lower efficacy in CD8(+) T cell priming and failed to protect the host once tumors had been established. Thus, specific in vivo targeting of DCs with coronavirus vectors in conjunction with appropriate conditioning of the microenvironment through Flt3L represents an efficient strategy for the generation of therapeutic antitumor immunity.http://europepmc.org/articles/PMC3842931?pdf=render
collection DOAJ
language English
format Article
sources DOAJ
author Christian Perez-Shibayama
Cristina Gil-Cruz
Monika Nussbacher
Eva Allgäuer
Luisa Cervantes-Barragan
Roland Züst
Burkhard Ludewig
spellingShingle Christian Perez-Shibayama
Cristina Gil-Cruz
Monika Nussbacher
Eva Allgäuer
Luisa Cervantes-Barragan
Roland Züst
Burkhard Ludewig
Dendritic cell-specific delivery of Flt3L by coronavirus vectors secures induction of therapeutic antitumor immunity.
PLoS ONE
author_facet Christian Perez-Shibayama
Cristina Gil-Cruz
Monika Nussbacher
Eva Allgäuer
Luisa Cervantes-Barragan
Roland Züst
Burkhard Ludewig
author_sort Christian Perez-Shibayama
title Dendritic cell-specific delivery of Flt3L by coronavirus vectors secures induction of therapeutic antitumor immunity.
title_short Dendritic cell-specific delivery of Flt3L by coronavirus vectors secures induction of therapeutic antitumor immunity.
title_full Dendritic cell-specific delivery of Flt3L by coronavirus vectors secures induction of therapeutic antitumor immunity.
title_fullStr Dendritic cell-specific delivery of Flt3L by coronavirus vectors secures induction of therapeutic antitumor immunity.
title_full_unstemmed Dendritic cell-specific delivery of Flt3L by coronavirus vectors secures induction of therapeutic antitumor immunity.
title_sort dendritic cell-specific delivery of flt3l by coronavirus vectors secures induction of therapeutic antitumor immunity.
publisher Public Library of Science (PLoS)
series PLoS ONE
issn 1932-6203
publishDate 2013-01-01
description Efficacy of antitumor vaccination depends to a large extent on antigen targeting to dendritic cells (DCs). Here, we assessed antitumor immunity induced by attenuated coronavirus vectors which exclusively target DCs in vivo and express either lymphocyte- or DC-activating cytokines in combination with a GFP-tagged model antigen. Tracking of in vivo transduced DCs revealed that vectors encoding for Fms-like tyrosine kinase 3 ligand (Flt3L) exhibited a higher capacity to induce DC maturation compared to vectors delivering IL-2 or IL-15. Moreover, Flt3L vectors more efficiently induced tumor-specific CD8(+) T cells, expanded the epitope repertoire, and provided both prophylactic and therapeutic tumor immunity. In contrast, IL-2- or IL-15-encoding vectors showed a substantially lower efficacy in CD8(+) T cell priming and failed to protect the host once tumors had been established. Thus, specific in vivo targeting of DCs with coronavirus vectors in conjunction with appropriate conditioning of the microenvironment through Flt3L represents an efficient strategy for the generation of therapeutic antitumor immunity.
url http://europepmc.org/articles/PMC3842931?pdf=render
work_keys_str_mv AT christianperezshibayama dendriticcellspecificdeliveryofflt3lbycoronavirusvectorssecuresinductionoftherapeuticantitumorimmunity
AT cristinagilcruz dendriticcellspecificdeliveryofflt3lbycoronavirusvectorssecuresinductionoftherapeuticantitumorimmunity
AT monikanussbacher dendriticcellspecificdeliveryofflt3lbycoronavirusvectorssecuresinductionoftherapeuticantitumorimmunity
AT evaallgauer dendriticcellspecificdeliveryofflt3lbycoronavirusvectorssecuresinductionoftherapeuticantitumorimmunity
AT luisacervantesbarragan dendriticcellspecificdeliveryofflt3lbycoronavirusvectorssecuresinductionoftherapeuticantitumorimmunity
AT rolandzust dendriticcellspecificdeliveryofflt3lbycoronavirusvectorssecuresinductionoftherapeuticantitumorimmunity
AT burkhardludewig dendriticcellspecificdeliveryofflt3lbycoronavirusvectorssecuresinductionoftherapeuticantitumorimmunity
_version_ 1725119857262854144